Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
Blood Adv ; 7(9): 1754-1761, 2023 05 09.
Article in English | MEDLINE | ID: mdl-36521029

ABSTRACT

Cancer cells take up amino acids from the extracellular space to drive cell proliferation and viability. Similar mechanisms are applied by immune cells, resulting in the competition between conventional T cells, or indeed chimeric antigen receptor (CAR) T cells and tumor cells, for the limited availability of amino acids within the environment. We demonstrate that T cells can be re-engineered to express SLC7A5 or SLC7A11 transmembrane amino acid transporters alongside CARs. Transporter modifications increase CAR T-cell proliferation under low tryptophan or cystine conditions with no loss of CAR cytotoxicity or increased exhaustion. Transcriptomic and phenotypic analysis reveals that downstream, SLC7A5/SLC7A11-modified CAR T cells upregulate intracellular arginase expression and activity. In turn, we engineer and phenotype a further generation of CAR T cells that express functional arginase 1/arginase 2 enzymes and have enhanced CAR T-cell proliferation and antitumor activity. Thus, CAR T cells can be adapted to the amino acid metabolic microenvironment of cancer, a hitherto recognized but unaddressed barrier for successful CAR T-cell therapy.


Subject(s)
Neoplasms , Receptors, Chimeric Antigen , Humans , T-Lymphocytes , Receptors, Chimeric Antigen/metabolism , Receptors, Antigen, T-Cell/genetics , Arginase/genetics , Arginase/metabolism , Large Neutral Amino Acid-Transporter 1/metabolism , Neoplasms/metabolism , Amino Acids/metabolism , Tumor Microenvironment
2.
Lancet Diabetes Endocrinol ; 10(8): 581-588, 2022 08.
Article in English | MEDLINE | ID: mdl-35780804

ABSTRACT

BACKGROUND: Radiotherapy is a valuable treatment in the management algorithm of pituitary adenomas and craniopharyngiomas. However, the risk of second brain tumour following radiotherapy is a major concern. We assessed this risk using non-irradiated patients with the same primary pathology and imaging surveillance as controls. METHODS: In this multicentre, retrospective cohort study, 4292 patients with pituitary adenoma or craniopharyngioma were identified from departmental registries at six adult endocrine centres (Birmingham, Oxford, Leeds, Leicester, and Bristol, UK and Ferrara, Italy). Patients with insufficient clinical data, known genetic predisposition to or history of brain tumour before study entry (n=532), and recipients of proton beam or stereotactic radiotherapy (n=81) were excluded. Data were analysed for 996 patients exposed to 2-dimensional radiotherapy, 3-dimensional conformal radiotherapy, or intensity-modulated radiotherapy, and compared with 2683 controls. FINDINGS: Over 45 246 patient-years, second brain tumours were reported in 61 patients (seven malignant [five radiotherapy, two controls], 54 benign [25 radiotherapy, 29 controls]). Radiotherapy exposure and older age at pituitary tumour detection were associated with increased risk of second brain tumour. Rate ratio for irradiated patients was 2·18 (95% CI 1·31-3·62, p<0·0001). Cumulative probability of second brain tumour was 4% for the irradiated and 2·1% for the controls at 20 years. INTERPRETATION: Irradiated adults with pituitary adenoma or craniopharyngioma are at increased risk of second brain tumours, although this risk is considerably lower than previously reported in studies using general population controls with no imaging surveillance. Our data clarify an important clinical question and guide clinicians when counselling patients with pituitary adenoma or craniopharyngioma on the risks and benefits of radiotherapy. FUNDING: Pfizer.


Subject(s)
Adenoma , Brain Neoplasms , Craniopharyngioma , Pituitary Neoplasms , Adenoma/diagnostic imaging , Adenoma/epidemiology , Adenoma/radiotherapy , Adult , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/epidemiology , Brain Neoplasms/radiotherapy , Cohort Studies , Craniopharyngioma/complications , Craniopharyngioma/diagnostic imaging , Craniopharyngioma/radiotherapy , Humans , Pituitary Neoplasms/diagnostic imaging , Pituitary Neoplasms/radiotherapy , Retrospective Studies
3.
Cancer Res ; 79(3): 611-624, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30545920

ABSTRACT

Neuroblastoma is the most common childhood solid tumor, yet the prognosis for high-risk disease remains poor. We demonstrate here that arginase 2 (ARG2) drives neuroblastoma cell proliferation via regulation of arginine metabolism. Targeting arginine metabolism, either by blocking cationic amino acid transporter 1 (CAT-1)-dependent arginine uptake in vitro or therapeutic depletion of arginine by pegylated recombinant arginase BCT-100, significantly delayed tumor development and prolonged murine survival. Tumor cells polarized infiltrating monocytes to an M1-macrophage phenotype, which released IL1ß and TNFα in a RAC-alpha serine/threonine-protein kinase (AKT)-dependent manner. IL1ß and TNFα established a feedback loop to upregulate ARG2 expression via p38 and extracellular regulated kinases 1/2 (ERK1/2) signaling in neuroblastoma and neural crest-derived cells. Proteomic analysis revealed that enrichment of IL1ß and TNFα in stage IV human tumor microenvironments was associated with a worse prognosis. These data thus describe an immune-metabolic regulatory loop between tumor cells and infiltrating myeloid cells regulating ARG2, which can be clinically exploited. SIGNIFICANCE: These findings illustrate that cross-talk between myeloid cells and tumor cells creates a metabolic regulatory loop that promotes neuroblastoma progression.


Subject(s)
Arginine/metabolism , Interleukin-1beta/metabolism , Macrophages/metabolism , Neuroblastoma/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Arginase/metabolism , Cell Line, Tumor , Humans , Interleukin-1beta/immunology , MAP Kinase Signaling System , Macrophages/immunology , Macrophages/pathology , Mice , Mice, Transgenic , Myeloid Cells/immunology , Myeloid Cells/metabolism , Myeloid Cells/pathology , Neuroblastoma/immunology , Neuroblastoma/pathology , Sarcoma, Ewing/immunology , Sarcoma, Ewing/metabolism , Sarcoma, Ewing/pathology , Tumor Microenvironment , Tumor Necrosis Factor-alpha/immunology
4.
Int J Cancer ; 142(7): 1490-1502, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29168171

ABSTRACT

Arginine is a semi-essential amino acid that plays a key role in cell survival and proliferation in normal and malignant cells. BCT-100, a pegylated (PEG) recombinant human arginase, can deplete arginine and starve malignant cells of the amino acid. Acute lymphoblastic leukemia (ALL) is the most common cancer of childhood, yet for patients with high risk or relapsed disease prognosis remains poor. We show that BCT-100 is cytotoxic to ALL blasts from patients in vitro by necrosis, and is synergistic in combination with dexamethasone. Against ALL xenografts, BCT-100 leads to a reduction in ALL engraftment and a prolongation of survival. ALL blasts express the arginine transporter CAT-1, yet the majority of blasts are arginine auxotrophic due to deficiency in either argininosuccinate synthase (ASS) or ornithine transcarbamylase (OTC). Although endogenous upregulation or retroviral transduced increases in ASS or OTC may promote ALL survival under moderately low arginine conditions, expression of these enzymes cannot prevent BCT-100 cytotoxicity at arginine depleting doses. RNA-sequencing of ALL blasts and supporting stromal cells treated with BCT-100 identifies a number of candidate pathways which are altered in the presence of arginine depletion. Therefore, BCT-100 provides a new clinically relevant therapeutic approach to target arginine metabolism in ALL.


Subject(s)
Antineoplastic Agents/pharmacology , Arginase/pharmacology , Arginine/metabolism , Metabolome/drug effects , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Recombinant Proteins/pharmacology , Animals , Cell Survival/drug effects , Dexamethasone/pharmacology , Drug Synergism , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Xenograft Model Antitumor Assays
5.
Oncotarget ; 8(38): 63506-63517, 2017 Sep 08.
Article in English | MEDLINE | ID: mdl-28969007

ABSTRACT

Paediatric sarcomas and brain tumours, remain cancers of significant unmet need, with a poor prognosis for patients with high risk disease or those who relapse, and significant morbidities from treatment for those that survive using standard treatment approaches. Novel treatment strategies, based on the underlying tumour biology, are needed to improve outcomes. Arginine is a semi-essential amino acid that is imported from the extracellular microenvironment or recycled from intracellular precursors through the combined expression of the enzymes ornithine transcarbamylase (OTC), argininosuccinate synthase (ASS) and argininosuccinate lyase (ASL) enzymes. The failure to express at least one of these recycling enzymes makes cells reliant on extracellular arginine - a state known as arginine auxotrophism. Here we show in large in silico patient cohorts that paediatric sarcomas and brain tumours express predominately the arginine transporter SLC7A1 and the arginine metabolising enzyme Arginase 2 (ARG2), but have low-absent expression of OTC. The arginine metabolic pathway correlated with the expression of genes associated with tumour pathogenesis, and overall survival in paediatric sarcomas. This gene signature of arginine auxotrophism indicates paediatric sarcomas and brain tumours are a viable target for therapeutic arginase drugs under current clinical trial development.

6.
Clin Teach ; 13(5): 388, 2016 10.
Article in English | MEDLINE | ID: mdl-27624201

Subject(s)
Diagnostic Errors , Humans
7.
Int J Cancer ; 139(3): 501-9, 2016 08 01.
Article in English | MEDLINE | ID: mdl-26913960

ABSTRACT

Renewed interest in the use of therapeutic enzymes combined with an improved knowledge of cancer cell metabolism, has led to the translation of several arginine depletion strategies into early phase clinical trials. Arginine auxotrophic tumors are reliant on extracellular arginine, due to the downregulation of arginosuccinate synthetase or ornithine transcarbamylase-key enzymes for intracellular arginine recycling. Engineered arginine catabolic enzymes such as recombinant human arginase (rh-Arg1-PEG) and arginine deiminase (ADI-PEG) have demonstrated cytotoxicity against arginine auxotrophic tumors. In this review, we discuss the molecular events triggered by extracellular arginine depletion that contribute to tumor cell death.


Subject(s)
Antineoplastic Agents/therapeutic use , Arginine/metabolism , Enzyme Therapy , Neoplasms/drug therapy , Neoplasms/metabolism , Animals , Antineoplastic Agents/pharmacology , Arginase/therapeutic use , Cell Proliferation , Cell Survival , Clinical Trials as Topic , Drug Evaluation, Preclinical , Enzyme Therapy/methods , Humans , Hydrolases/therapeutic use , Metabolic Networks and Pathways/drug effects , Neoplasms/enzymology , Signal Transduction/drug effects , Stress, Physiological/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...